Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Front Immunol ; 11: 476, 2020.
Article in English | MEDLINE | ID: mdl-32265927

ABSTRACT

Replacement therapy with coagulation factor VIII (FVIII) represents the current clinical treatment for patients affected by hemophilia A (HA). This treatment while effective is, however, hampered by the formation of antibodies which inhibit the activity of infused FVIII in up to 30% of treated patients. Immune tolerance induction (ITI) protocols, which envisage frequent infusions of high doses of FVIII to confront this side effect, dramatically increase the already high costs associated to a patient's therapy and are not always effective in all treated patients. Therefore, there are clear unmet needs that must be addressed in order to improve the outcome of these treatments for HA patients. Taking advantage of preclinical mouse models of hemophilia, several strategies have been proposed in recent years to prevent inhibitor formation and eradicate the pre-existing immunity to FVIII inhibitor positive patients. Herein, we will review some of the most promising strategies developed to avoid and eradicate inhibitors, including the use of immunomodulatory drugs or molecules, oral or transplacental delivery as well as cell and gene therapy approaches. The goal is to improve and potentiate the current ITI protocols and eventually make them obsolete.


Subject(s)
Factor VIII/immunology , Hemophilia A/drug therapy , Immune Tolerance/drug effects , Immunologic Factors/therapeutic use , Isoantibodies/immunology , Adoptive Transfer , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Drug Carriers , Drug Compounding , Drug Evaluation, Preclinical , Factor IXa/immunology , Factor VIII/administration & dosage , Factor VIII/therapeutic use , Factor X/immunology , Female , Fetal Therapies , Genetic Therapy , Hemophilia A/immunology , Hemophilia A/therapy , Humans , Immunoglobulin Fc Fragments/administration & dosage , Immunotherapy, Adoptive , Isoantibodies/biosynthesis , Lymphoid Tissue/immunology , Mice , Models, Animal , Plant Cells , Pregnancy , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes, Regulatory/transplantation
2.
Int J Hematol ; 111(1): 20-30, 2020 Jan.
Article in English | MEDLINE | ID: mdl-30350119

ABSTRACT

Hemophilia A is a congenital disorder caused by deficiency or malfunction of coagulation factor (F) VIII. While exogenously provided FVIII effectively reduces bleeding complications in many hemophilia A patients, multiple efforts are underway to develop new drugs to meet the needs that conventional FVIII agents do not. We have been long engaged in creating and clinically developing a humanized anti-FIXa/FX asymmetric bispecific IgG antibody with a FVIIIa-cofactor activity. Since this project was born from a creative and unique idea, our group recognized from the first that it would face many difficulties in the course of research including establishment of industrial manufacturability of an asymmetric bispecific IgG antibody. The group actually faced various challenges, but addressed all of them during about 10 years of research, and successfully created the potent humanized bispecific antibody, emicizumab. Emicizumab has showed clinical benefits in the human trials among which the first one was started in 2012, and has been currently approved in US, EU, Japan, and some other countries. It is now expected to improve the quality of life of patients and their families. In this article, we review the course of the research and clinical development of emicizumab, and describe its molecular characteristics.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Factor IXa/immunology , Factor VIIIa/metabolism , Factor X/immunology , Hemophilia A/drug therapy , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Blood Coagulation/drug effects , Factor VIII/immunology , Factor VIII/metabolism , Hemophilia A/immunology , Humans , Protein Engineering
3.
Pediatr Blood Cancer ; 66(11): e27886, 2019 11.
Article in English | MEDLINE | ID: mdl-31348595

ABSTRACT

BACKGROUND: Emicizumab is a bispecific antibody that bridges factor IXa and factor X to restore hemostasis in patients with hemophilia A (HA). Its efficacy and safety have been proven in multicenter trials. However, real world data regarding its use in very young children are currently lacking. Ancillary test results for monitoring emicizumab's hemostatic effect and their clinical correlations are scarce. METHODS: Children with HA and inhibitors treated by emicizumab were prospectively followed at our center. Laboratory follow-up included rotational thromboelastometry (ROTEM) and thrombin generation (TG), prior to and during treatment. RESULTS: Eleven children whose median age was 26 months were treated by emicizumab and followed for a median of 36 weeks. During follow-up, none experienced hemarthrosis or any other spontaneous bleeds. For 7/11 patients, emicizumab prophylaxis was sufficient to maintain hemostasis without additional supplemental therapy. Only 4/11 patients were occasionally treated with recombinant activated FVII for trauma. Two minor surgeries were safely performed without supplemental therapy while another procedure was complicated by major bleeding. TG parameters improved for all patients, correlating with their clinical status. Interestingly, the lowest TG values were obtained for patients experiencing bleeding episodes, while ROTEM parameters in all patients were close to the normal range. CONCLUSIONS: This study confirms the safety and efficacy of emicizumab in reducing bleeds in young children with HA with inhibitors, including infants. However, surgeries warrant caution as emicizumab prophylaxis may not be sufficient for some procedures. TG may more accurately reflect the hemostasis state than ROTEM in pediatric patients treated with emicizumab.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Hemophilia A/drug therapy , Hemorrhage/prevention & control , Blood Loss, Surgical/prevention & control , Child, Preschool , Factor IXa/immunology , Factor X/immunology , Follow-Up Studies , Hemophilia A/blood , Hemophilia A/complications , Hemorrhage/etiology , Humans , Infant , Male , Prospective Studies , Thrombelastography , Thrombin/analysis , Treatment Outcome
4.
J Thromb Haemost ; 16(7): 1383-1390, 2018 07.
Article in English | MEDLINE | ID: mdl-29734520

ABSTRACT

Essentials Emicizumab (Emi) affects the APTT-based assays of factor (F)VIII activity and inhibitor titer. A mixture of two anti-Emi monoclonal antibodies (mAb) effectively neutralized the Emi activity. Anti-Emi mAbs completely eliminated the influence of Emi on FVIII activity and inhibitor titer. The inclusion of anti-Emi mAbs in routine FVIII assays would be useful for Emi-treated patients. SUMMARY: Background Emicizumab is an anti-factor (F)IXa/X bispecific monoclonal antibody (mAb), mimicking the factor (F)VIIIa cofactor activity. Emicizumab does not require activation by thrombin and its shortening effect on the activated partial prothrombin time (APTT) is more pronounced than that of factor (F)VIII. APTT-based FVIII activity (FVIII:C) and FVIII inhibiter titer measurements are influenced by the presence of emicizumab. Aim To establish a reliable APTT-based assay to measure FVIII in the presence of emicizumab. Methods Plasmas from hemophilia A (HA) patients without or with inhibitors were studied using one-stage FVIII:C and Bethesda inhibitor assays. Two recombinant anti-idiotype mAbs to emicizumab (anti-emicizumab mAbs) were prepared, rcAQ8 to anti-FIXa-Fab and rcAJ540 to anti-FX-Fab. Results The combined anti-idiotype mAbs (2000 nm each) eliminated the effects of emicizumab on APTTs of HA plasmas without or with inhibitor by competitive inhibition of antibody binding to FIX(a)/FX(a). Measurements of FVIII coagulation activity in HA plasmas without inhibitor were overestimated in the presence of emicizumab (1 µm = ~150 µg mL-1 ) at all reference levels of FVIII. The addition of anti-emicizumab mAbs to the assay mixtures completely neutralized the emicizumab and facilitated accurate determination of FVIII:C. Anti-FVIII inhibitor titers were undetectable in the presence of emicizumab in HA plasmas with inhibitor or normal plasmas mixed with anti-FVIII neutralizing antibodies. These effects of emicizumab were completely counteracted by the addition of the anti-idiotype mAbs, allowing accurate assessment of inhibitor titers. Conclusion The in vitro inclusion of anti-emicizumab mAbs in the standard one-stage coagulation assays prevented interference by emicizumab and enabled accurate measurements of FVIII:C and inhibitor titers.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Neutralizing/blood , Blood Coagulation/drug effects , Coagulants/pharmacology , Factor VIII/analysis , Hemophilia A/blood , Partial Thromboplastin Time , Antibodies, Bispecific/blood , Antibodies, Bispecific/immunology , Antibodies, Monoclonal, Humanized/blood , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Neutralizing/immunology , Binding, Competitive , Coagulants/blood , Coagulants/immunology , Dose-Response Relationship, Drug , Factor IXa/immunology , Factor IXa/metabolism , Factor VIII/immunology , Factor Xa/immunology , Factor Xa/metabolism , Hemophilia A/diagnosis , Hemophilia A/immunology , Humans , Predictive Value of Tests , Protein Binding , Reproducibility of Results
5.
Yakugaku Zasshi ; 137(7): 831-836, 2017.
Article in Japanese | MEDLINE | ID: mdl-28674296

ABSTRACT

Nearly fifty monoclonal antibodies have been approved to date, and the market for monoclonal antibodies is expected to continue to grow. Since global competition in the field of antibody therapeutics is intense, we need to establish novel antibody engineering technologies to provide true benefit for patients, with differentiated product values. Bispecific antibodies are among the next generation of antibody therapeutics that can bind to two different target antigens by the two arms of immunoglobulin G (IgG) molecule, and are thus believed to be applicable to various therapeutic needs. Until recently, large scale manufacturing of human IgG bispecific antibody was impossible. We have established a technology, named asymmetric re-engineering technology (ART)-Ig, to enable large scale manufacturing of bispecific antibodies. Three examples of next generation antibody therapeutics using ART-Ig technology are described. Recent updates on bispecific antibodies against factor IXa and factor X for the treatment of hemophilia A, bispecific antibodies against a tumor specific antigen and T cell surface marker CD3 for cancer immunotherapy, and bispecific antibodies against two different epitopes of soluble antigen with pH-dependent binding property for the elimination of soluble antigen from plasma are also described.


Subject(s)
Antibodies, Bispecific , Antibodies, Monoclonal , Immunoglobulin G , Protein Engineering/methods , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , CD3 Complex/immunology , Epitopes/immunology , Factor IXa/immunology , Factor X/immunology , Hemophilia A/therapy , Humans , Hydrogen-Ion Concentration , Immunotherapy , Neoplasms/therapy , Protein Binding , Solubility
6.
Thromb Haemost ; 117(7): 1348-1357, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28451690

ABSTRACT

Emicizumab, a humanised bispecific antibody recognising factors (F) IX/IXa and X/Xa, can accelerate FIXa-catalysed FX activation by bridging FIXa and FX in a manner similar to FVIIIa. However, details of the emicizumab-antigen interactions have not been reported so far. In this study, we first showed by surface plasmon resonance analysis that emicizumab bound FIX, FIXa, FX, and FXa with moderate affinities (KD = 1.58, 1.52, 1.85, and 0.978 µM, respectively). We next showed by immunoblotting analysis that emicizumab recognised the antigens' epidermal growth factor (EGF)-like domains. We then performed KD-based simulation of equilibrium states in plasma for quantitatively predicting the ways that emicizumab would interact with the antigens. The simulation predicted that only a small part of plasma FIX, FX, and emicizumab would form antigen-bridging FIX-emicizumab-FX ternary complex, of which concentration would form a bell-shaped relationship with emicizumab concentration. The bell-shaped concentration dependency was reproduced by plasma thrombin generation assays, suggesting that the plasma concentration of the ternary complex would correlate with emicizumab's cofactor activity. The simulation also predicted that at 10.0-100 µg/ml of emicizumab-levels shown in a previous study to be clinically effective-the majority of plasma FIX, FX, and emicizumab would exist as monomers. In conclusion, emicizumab binds FIX/FIXa and FX/FXa with micromolar affinities at their EGF-like domains. The KD-based simulation predicted that the antigen-bridging ternary complex formed in circulating plasma would correlate with emicizumab's cofactor activity, and the majority of FIX and FX would be free and available for other coagulation reactions.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Factor VIIIa/immunology , Antibodies, Bispecific/blood , Antibodies, Monoclonal, Humanized/blood , Antibody Specificity , Antigen-Antibody Reactions , Binding Sites , Biomimetic Materials/pharmacology , Computer Simulation , Factor IX/antagonists & inhibitors , Factor IX/immunology , Factor IXa/antagonists & inhibitors , Factor IXa/immunology , Factor X/antagonists & inhibitors , Factor X/immunology , Factor Xa/immunology , Factor Xa Inhibitors/blood , Factor Xa Inhibitors/immunology , Factor Xa Inhibitors/pharmacology , Humans , Models, Immunological
7.
Thromb Res ; 141 Suppl 2: S34-5, 2016 May.
Article in English | MEDLINE | ID: mdl-27207420

ABSTRACT

There are some issues in the current factor (F)VIII replacement therapy for severe hemophilia A. One is mental and physical burden for the multiple intravenous infusions, and the other is difficulty in the hemostatic treatment for the patients with FVIII inhibitor. The development of novel drug with fully hemostatic effect, simply procedure, and long-acting reaction has been expected. Recently, FVIIIa-mimicking humanized recombinant bispecific antibody (ACE910) against FIXa and FX was developed. In the non-human clinical study, primate model of acquired hemophilia A demonstrated that the ACE910 was effective on both on-going and spontaneous bleedings. A phase I clinical study was conducted in healthy adults by single subcutaneous infusion of ACE910, followed by the patients' part study, Japanese patients with severe hemophilia A without or with inhibitor were treated with once-weekly subcutaneous injection of ACE910 at three dose levels for 12 successive weeks. There was no significant adverse event related to ACE910 in the clinical and laboratorial findings, and t1/2 of ACE910 was ∼30 days. The median annual bleeding rates were reduced very markedly dose-dependently, independently of inhibitor. Furthermore, among the patients with dose escalation, bleeding rate was decreased as ACE910 dose was increased. In conclusion, ACE910 would have a number of promising features: its high subcutaneous bioavailability and long half-life make the patients possible to be injected subcutaneously with a once-a-week or less frequency. In addition, ACE910 would provide the bleeding prophylactic efficacy, independently of inhibitor.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Factor VIII/metabolism , Hemophilia A/drug therapy , Hemophilia A/metabolism , Hemostatics/therapeutic use , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Clinical Trials as Topic , Factor IXa/immunology , Factor IXa/metabolism , Factor X/immunology , Factor X/metabolism , Hemophilia A/blood , Hemophilia A/immunology , Hemorrhage/blood , Hemorrhage/immunology , Hemorrhage/metabolism , Hemorrhage/prevention & control , Hemostatics/immunology , Hemostatics/pharmacology , Humans
8.
Rinsho Ketsueki ; 56(6): 623-31, 2015 Jun.
Article in Japanese | MEDLINE | ID: mdl-26256871

ABSTRACT

Unmet needs of current hemophilia A treatment include the requirement for frequent intravenous infusions, inhibitor development, and containment of high medical costs. In order to overcome these issues, we produced FVIII which mimics a bispecific antibody against FIXa/FX. ACE910 demonstrated hemostatic effects on both ongoing and spontaneous joint bleeding in the primate acquired hemophilia A model. Recently, a phase 1 study for PK, PD, and the safety of ACE910 was initiated. The t1/2 was approximately 30 days. There were no severe ACE910 related adverse events. Furthermore, bleeding was remarkably decreased by weekly subcutaneous administration in patients with severe hemophilia A, regardless of whether an inhibitor was used. ACE910 has the remarkable advantages of prophylactic efficacy which can be achieved by convenient subcutaneous administrations at a markedly reduced frequency.


Subject(s)
Antibodies/therapeutic use , Factor IXa/immunology , Factor X/immunology , Hemophilia A/drug therapy , Hemophilia B/drug therapy , Animals , Clinical Trials as Topic , Hemophilia A/immunology , Hemophilia B/immunology , Humans
9.
Blood ; 124(20): 3165-71, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25274508

ABSTRACT

ACE910 is a humanized anti-factor IXa/X bispecific antibody mimicking the function of factor VIII (FVIII). We previously demonstrated in nonhuman primates that a single IV dose of ACE910 exerted hemostatic activity against hemophilic bleeds artificially induced in muscles and subcutis, and that a subcutaneous (SC) dose of ACE910 showed a 3-week half-life and nearly 100% bioavailability, offering support for effective prophylaxis for hemophilia A by user-friendly SC dosing. However, there was no direct evidence that such SC dosing of ACE910 would prevent spontaneous bleeds occurring in daily life. In this study, we newly established a long-term primate model of acquired hemophilia A by multiple IV injections of an anti-primate FVIII neutralizing antibody engineered in mouse-monkey chimeric form to reduce its antigenicity. The monkeys in the control group exhibited various spontaneous bleeding symptoms as well as continuous prolongation of activated partial thromboplastin time; notably, all exhibited joint bleeds, which are a hallmark of hemophilia. Weekly SC doses of ACE910 (initial 3.97 mg/kg followed by 1 mg/kg) significantly prevented these bleeding symptoms; notably, no joint bleeding symptoms were observed. ACE910 is expected to prevent spontaneous bleeds and joint damage in hemophilia A patients even with weekly SC dosing, although appropriate clinical investigation is required.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Factor IXa/immunology , Factor X/immunology , Hemophilia A/complications , Hemorrhage/complications , Hemorrhage/prevention & control , Animals , Antibodies, Bispecific/immunology , Antibodies, Monoclonal, Humanized/immunology , Cell Line , Disease Models, Animal , Hemorrhage/pathology , Humans , Joints/drug effects , Joints/pathology , Macaca fascicularis , Male , Mice
10.
J Thromb Haemost ; 12(2): 206-13, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24738137

ABSTRACT

BACKGROUND: We previously reported that a humanized anti-factor IXa/X bispecific antibody, hBS23, mimics the function of FVIII even in the presence of FVIII inhibitors, and has preventive hemostatic activity against bleeding in an animal model of acquired hemophilia A. After further molecular engineering of hBS23, we recently identified an improved humanized bispecific antibody, ACE910, for clinical investigation. OBJECTIVES: To elucidate the in vivo hemostatic potency of ACE910 by examining its effect against ongoing bleeds, and to determine its pharmacokinetic parameters for discussion of its potency for prophylactic use. METHODS: A nonhuman primate model of acquired hemophilia A was established by injecting anti-primate FVIII neutralizing antibody. When bleeds emerged following an artificial bleed-inducing procedure, either ACE910 or recombinant porcine FVIII (rpoFVIII) was intravenously administered. rpoFVIII was additionally administered twice daily on the following 2 days. Bleeding symptoms were monitored for 3 days. A pharmacokinetic study and multiple-dosing simulations of ACE910 were also performed. RESULTS: A single bolus of 1 or 3 mg kg⁻¹ ACE910 showed hemostatic activity comparable to that of 10 U kg⁻¹ (twice daily) rpoFVIII against ongoing bleeds. The determined ACE910 pharmacokinetic parameters included a long half-life (3 weeks) and high subcutaneous bioavailability (nearly 100%). The simulation results based on pharmacokinetic parameters indicated that the above hemostatic level could be maintained with once-weekly subcutaneous administration of ACE910, suggesting the possibility of more effective prophylaxis. CONCLUSIONS: ACE910 may offer an alternative on-demand treatment option for patients with hemophilia A, as well as user-friendly and aggressive routine supplementation.


Subject(s)
Antibodies/immunology , Factor IXa/immunology , Factor X/immunology , Hemophilia A/therapy , Hemostasis/immunology , Animals , CHO Cells , Cricetinae , Cricetulus , Cross Reactions , Disease Models, Animal , HEK293 Cells , Humans , Macaca fascicularis , Male
11.
Nat Med ; 18(10): 1570-4, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23023498

ABSTRACT

Hemophilia A is a bleeding disorder resulting from coagulation factor VIII (FVIII) deficiency. Exogenously provided FVIII effectively reduces bleeding complications in patients with severe hemophilia A. In approximately 30% of such patients, however, the 'foreignness' of the FVIII molecule causes them to develop inhibitory antibodies against FVIII (inhibitors), precluding FVIII treatment in this set of patients. Moreover, the poor pharmacokinetics of FVIII, attributed to low subcutaneous bioavailability and a short half-life of 0.5 d, necessitates frequent intravenous injections. To overcome these drawbacks, we generated a humanized bispecific antibody to factor IXa (FIXa) and factor X (FX), termed hBS23, that places these two factors into spatially appropriate positions and mimics the cofactor function of FVIII. hBS23 exerted coagulation activity in FVIII-deficient plasma, even in the presence of inhibitors, and showed in vivo hemostatic activity in a nonhuman primate model of acquired hemophilia A. Notably, hBS23 had high subcutaneous bioavailability and a 2-week half-life and would not be expected to elicit the development of FVIII-specific inhibitory antibodies, as its molecular structure, and hence antigenicity, differs from that of FVIII. A long-acting, subcutaneously injectable agent that is unaffected by the presence of inhibitors could markedly reduce the burden of care for the treatment of hemophilia A.


Subject(s)
Antibodies, Bispecific , Factor IXa/immunology , Factor VIII/physiology , Factor X/immunology , Hemophilia A/therapy , Hemostasis , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Hemophilia A/immunology , Macaca fascicularis
13.
J Immunol ; 182(3): 1674-80, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19155517

ABSTRACT

We previously reported that some human antiphospholipid Abs (aPL) in patients with the antiphospholipid syndrome (APS) bind to the homologous enzymatic domains of thrombin and the activated coagulation factor X (FXa). Moreover, some of the reactive Abs are prothrombotic and interfere with inactivation of thrombin and FXa by antithrombin (AT). Considering the enzymatic domain of activated coagulation factor IX (FIXa) is homologous to those of thrombin and FXa, we hypothesized that some aPLs in APS bind to FIXa and hinder AT inactivation of FIXa. To test this hypothesis, we searched for IgG anti-FIXa Abs in APS patients. Once the concerned Abs were found, we studied the effects of the Ab on FIXa inactivation by AT. We found that 10 of 12 patient-derived monoclonal IgG aPLs bound to FIXa and that IgG anti-FIXa Abs in APS patients were significantly higher than those in normal controls (p < 0.0001). Using the mean + 3 SD of 30 normal controls as the cutoff, the IgG anti-FIXa Abs were present in 11 of 38 (28.9%) APS patients. Importantly, 4 of 10 FIXa-reactive monoclonal aPLs (including the B2 mAb generated against beta(2)-glycoprotein I significantly hindered AT inactivation of FIXa. More importantly, IgG from two positive plasma samples were found to interfere with AT inactivation of FIXa. In conclusion, IgG anti-FIXa Ab occurred in approximately 30% of APS patients and could interfere with AT inactivation of FIXa. Because FIXa is an upstream procoagulant factor, impaired AT regulation of FIXa might contribute more toward thrombosis than the dysregulation of the downstream FXa and thrombin.


Subject(s)
Antibodies, Antiphospholipid/physiology , Antiphospholipid Syndrome/immunology , Antithrombins/physiology , Factor IXa/immunology , Factor IXa/metabolism , Adolescent , Adult , Aged , Antibodies, Antiphospholipid/blood , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/physiology , Antiphospholipid Syndrome/blood , Antithrombins/metabolism , Binding Sites, Antibody , Factor IXa/antagonists & inhibitors , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/physiology , Male , Middle Aged , Protein Binding/immunology
14.
Haemophilia ; 14 Suppl 3: 138-46, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18510534

ABSTRACT

In mild/moderate haemophilia A (MHA) patients, many factor VIII (FVIII) gene defects, mainly missense mutations, have been identified and greatly improved the understanding of the structure and function of FVIII molecule. Characterization of the molecular mechanisms involved in MHA has helped to identify regions critical for proper FVIII biosynthesis, thrombin activation, intramolecular stability as well as binding regions for important intermolecular interactions with von Willebrand factor, factor IXa and the phospholipid surface. Some missense mutations were also recognized as contributing factors to inhibitor development in MHA, in parallel to acquired factors such as inflammatory state or intensity of treatment. Treatment of MHA with inhibitor patients raises questions on how best to stop or prevent bleeding episodes and eradicate the inhibitor. Longitudinal data collection is currently being conducted in France and Belgium to enhance our knowledge in this field and to further help make treatment decision. The description of mutations in MHA finally contributed to the identification of epitopes involved in the immune response to FVIII. In some patients, the epitope specificity of inhibitor antibodies recognizing normal exogenous FVIII alone and not patient ('self') FVIII was described. This distinguished epitope specificity could also be demonstrated at the T-cell clonal level. One might expect that these molecular studies will have a major impact on development of new FVIII products in the future.


Subject(s)
Epitopes/immunology , Factor VIII/immunology , Hemophilia A/immunology , Immune Tolerance/immunology , Mutation, Missense/immunology , Antibodies/immunology , Deamino Arginine Vasopressin/therapeutic use , Factor IXa/immunology , Factor VIII/genetics , Genotype , Hemophilia A/drug therapy , Hemophilia A/genetics , Hemorrhage/drug therapy , Hemostatics/therapeutic use , Humans , Mutation, Missense/genetics , Thrombin/immunology
15.
J Thromb Haemost ; 6(2): 315-22, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18088341

ABSTRACT

BACKGROUND: Factor VIIIa (FVIIIa) binds to activated FIX and enhances the activation of FX by several orders of magnitude. Deficiency of FVIII causes the bleeding disorder hemophilia A and is treated by i.v. infusion of FVIII concentrates. OBJECTIVES: To explore whether or not FVIII activity can be supplied by alternative molecules, e.g. molecules with FIXa-binding activity. METHODS: Conventional hybdridoma technology was used to discover antibodies exhibiting FVIII-like activity. RESULTS: We identified a series of antibodies specific for human FIX that mimicked the stimulatory effect of FVIIIa on FIXa. Upon binding to human FIXa, these antibodies enhanced the protease activity of FIXa towards its natural substrate FX about tenfold. A similar enhancement was also achieved with 5 pm FVIIIa (i.e. 16 mU mL(-1) or 1.6% activated FVIII). Procoagulant activity of these anti-FIXa antibodies was observed in model systems containing purified proteins as well as in plasma. CONCLUSION: Our findings show that FVIII can, at least partially, be replaced by an unrelated molecule. Procoagulant antibodies might potentially aid the development of an FVIII substitute for hemophilia A treatment.


Subject(s)
Antibodies, Monoclonal/pharmacology , Factor IX/immunology , Factor IXa/agonists , Animals , Antibodies, Monoclonal/immunology , Cell-Free System , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Factor IXa/immunology , Factor VIIIa/physiology , Factor X/metabolism , Hemophilia A/drug therapy , Humans , Hybridomas/immunology , Male , Mice , Mice, Inbred BALB C
16.
Arterioscler Thromb Vasc Biol ; 27(4): 722-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17272750

ABSTRACT

Currently available anticoagulants are limited by modest therapeutic benefits, narrow clinical applications, increased bleeding risk, and drug-induced thrombophilia. Because factor IX plays a pivotal role in tissue factor (TF)-mediated thrombin generation, it may represent a promising target for drug development. Several methods of attenuating factor IX activity, including monoclonal antibodies, synthetic active site-blocked competitive inhibitors, oral inhibitors, and RNA aptamers, have undergone investigation. This review summarizes present knowledge of factor IX inhibitors with emphasis on biology, pharmacology, preclinical data, and early-phase clinical experience in humans.


Subject(s)
Anticoagulants/pharmacology , Factor IXa/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Aptamers, Nucleotide/pharmacology , Blood Coagulation/physiology , Factor IX/chemistry , Factor IX/physiology , Factor IXa/immunology , Humans , Structure-Activity Relationship
18.
J Biol Chem ; 278(33): 31297-302, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12782629

ABSTRACT

During blood coagulation factor IXa binds to factor VIIIa on phospholipid membranes to form an enzymatic complex, the tenase complex. To test whether there is a protein-protein contact site between the gamma-carboxyglutamic acid (Gla) domain of factor IXa and factor VIIIa, we demonstrated that an antibody to the Gla domain of factor IXa inhibited factor VIIIa-dependent factor IXa activity, suggesting an interaction of the factor IXa Gla domain with factor VIIIa. To study this interaction, we synthesized three analogs of the factor IXa Gla domain (FIX1-47) with Phe-9, Phe-25, or Val-46 replaced, respectively, with benzoylphenylalanine (BPA), a photoactivatable cross-linking reagent. These factor IX Gla domain analogs maintain native tertiary structure, as demonstrated by calcium-induced fluorescence quenching and phospholipid binding studies. In the absence of phospholipid membranes, FIX1-47 was able to inhibit factor IXa activity. This inhibition is dependent on the presence of factor VIIIa, suggesting a contact site between the factor IXa Gla domain and factor VIIIa. To demonstrate a direct interaction we did cross-linking experiments with FIX1-479BPA, FIX1-4725BPA, and FIX1-4746BPA. Covalent cross-linking to factor VIIIa was observed primarily with FIX1-4725BPA and to a much lesser degree with FIX1-4746BPA. Immunoprecipitation experiments with an antibody to the C2 domain of factor VIIIa indicate that the factor IX Gla domain cross-links to the A3-C1-C2 domain of factor VIIIa. These results suggest that the factor IXa Gla domain contacts factor VIIIa in the tenase complex through a contact site that includes phenylalanine 25 and perhaps valine 46.


Subject(s)
Cysteine Endopeptidases/metabolism , Factor IXa/metabolism , Factor VIIIa/metabolism , Neoplasm Proteins/metabolism , Phenylalanine/analogs & derivatives , Amino Acid Sequence , Amino Acid Substitution , Antibodies/pharmacology , Cross-Linking Reagents/pharmacology , Factor IXa/chemistry , Factor IXa/immunology , Humans , Molecular Sequence Data , Phenylalanine/metabolism , Phenylalanine/pharmacology , Protein Structure, Tertiary , Valine/metabolism
19.
Arterioscler Thromb Vasc Biol ; 22(3): 517-22, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11884300

ABSTRACT

10C12, a human antibody F(ab')2, which specifically binds to the gamma-carboxyglutamic acid domain of factor IX/factor IXa (F.IX/IXa), interferes with all known coagulation processes in which F.IX/IXa is involved. In a rabbit model of carotid artery injury, intravenous administration of 10C12 or heparin decreased thrombosis dose dependently. The dose that resulted in a 90% reduction of thrombus mass (ED90) was a 30-microg/kg bolus of 10C12 or a 100-U/kg bolus plus 1.0 U x kg(-1) x min(-1) infusion of heparin. Heparin, at and below the ED90, significantly prolonged coagulation times and cuticle bleeding times. In contrast, 10C12 had no effect on coagulation or bleeding times at doses up to 4 times the ED90. To further evaluate the effect of 10C12 on bleeding, it was compared with heparin in a novel model of blood loss. At the ED90 of heparin, blood loss induced by a standardized injury to the vasculature of the rabbit tibia increased to more than 2 times that of saline controls. In contrast, the dose of 10C12 required to produce a similar increase in blood loss was more than 30 times the ED90. The antithrombotic potency and relative safety of this fully human antibody suggests that it may have therapeutic value for treatment of thrombotic disorders.


Subject(s)
Anticoagulants/therapeutic use , Carotid Artery Thrombosis/prevention & control , Factor IX/antagonists & inhibitors , Factor IXa/antagonists & inhibitors , Immunoglobulin Fab Fragments/therapeutic use , Animals , Anticoagulants/adverse effects , Anticoagulants/pharmacology , Blood Coagulation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Factor IX/immunology , Factor IXa/immunology , Hemorrhage/etiology , Heparin/adverse effects , Heparin/pharmacology , Heparin/therapeutic use , Humans , Immunoglobulin Fab Fragments/adverse effects , Immunoglobulin Fab Fragments/pharmacology , Kinetics , Rabbits , Reproducibility of Results , Tibia/blood supply
20.
Nihon Rinsho ; 60(3): 525-30, 2002 Mar.
Article in Japanese | MEDLINE | ID: mdl-11904968

ABSTRACT

Monoclonal antibody therapies have conducted to not only hematologic malignancies but also disorders of hemostasis and coagulation. This article describes the recent advances of monoclonal antibody therapy for bleeding disorders such as idiopathic thrombocytopenic purpura(ITP), hemophilia A, disseminated intravascular coagulation(DIC), and thrombosis. Rituximab, chimeric anti-CD20 monoclonal antibody treatment has a valuable effect in the patients with ITP, and clinical trials using anti-CD40 ligand monoclonal antibody for ITP are underway. Anti-CD40 ligand monoclonal antibody can be an alternative therapy for hemophilia A patients with inhibitors to factor VIII. In thrombosis, anti-tissue factor monoclonal antibody and anti-factor IX(a) monoclonal antibody were established as novel anticoagulant regents. Plasminogen activator inhibitor-1(PAI-1) increases in endotoxin-induced DIC and many thrombotic diseases such as myocardial infarction, type 2 diabetes mellitus, and hyperlipidemia. Anti-PAI-1 monoclonal antibody reduced fibrin deposition in DIC mouse model. Treatment of these monoclonal antibodies for the molecules regulating coagulation-fibrinolysis system may be utilized for acute coronary syndrome and venous thrombosis.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Blood Coagulation Disorders/therapy , Animals , Antibodies, Monoclonal, Murine-Derived , Antigens, CD20/immunology , CD40 Ligand/immunology , Clinical Trials as Topic , Factor IXa/immunology , Humans , Mice , Plasminogen Activator Inhibitor 1/immunology , Rituximab
SELECTION OF CITATIONS
SEARCH DETAIL
...